Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Joint single-cell measurements of nuclear proteins and RNA in vivo

Abstract

Identifying gene-regulatory targets of nuclear proteins in tissues is a challenge. Here we describe intranuclear cellular indexing of transcriptomes and epitopes (inCITE-seq), a scalable method that measures multiplexed intranuclear protein levels and the transcriptome in parallel across thousands of nuclei, enabling joint analysis of transcription factor (TF) levels and gene expression in vivo. We apply inCITE-seq to characterize cell state-related changes upon pharmacological induction of neuronal activity in the mouse brain. Modeling gene expression as a linear combination of quantitative protein levels revealed genome-wide associations of each TF and recovered known gene targets. TF-associated genes were coexpressed as distinct modules that each reflected positive or negative TF levels, showing that our approach can disentangle relative putative contributions of TFs to gene expression and add interpretability to inferred gene networks. inCITE-seq can illuminate how combinations of nuclear proteins shape gene expression in native tissue contexts, with direct applications to solid or frozen tissues and clinical specimens.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: inCITE-seq simultaneously measures intranuclear protein and RNA levels at single-nucleus resolution.
Fig. 2: In vivo application of inCITE-seq shows cell type-specific protein expression in the mouse hippocampus.
Fig. 3: inCITE-seq measures changes in nuclear TF levels after stimulation of the mouse hippocampus.
Fig. 4: Inferring TF effects on gene and module expression using joint protein and transcriptome measurements.

Similar content being viewed by others

Data availability

Raw gene expression count matrices of all inCITE-seq data, BAM files of mapped reads and the matrix of mouse hippocampus inCITE-seq data jointly embedded with snRNA-seq data are available on Gene Expression Omnibus under the accession GSE163480. Data from Habib et al.54 are available under GSE143758. Data from MULTI-seq used to compare RNA complexity in HEK cells are available under GSE129578. Databases of TF motifs (CIS-BS and JASPAR2018_CORE_vertebrates_non-redundant) are available at http://cisbp.ccbr.utoronto.ca and http://jaspar2018.genereg.net, respectively. Source data are provided with this paper.

Code availability

Code used for analyses is available at https://github.com/klarman-cell-observatory/inCITE-seq.

References

  1. Habib, N. et al. Div-seq: single-nucleus RNA-seq reveals dynamics of rare adult newborn neurons. Science 353, 925–928 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Slyper, M. et al. A single-cell and single-nucleus RNA-seq toolbox for fresh and frozen human tumors. Nat. Med. 26, 792–802 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lacar, B. et al. Nuclear RNA-seq of single neurons reveals molecular signatures of activation. Nat. Commun. 7, 11022 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. van den Brink, S. C. et al. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Methods 14, 935–936 (2017).

    Article  PubMed  CAS  Google Scholar 

  6. Drokhlyansky, E. et al. The human and mouse enteric nervous system at single-cell resolution. Cell 182, 1606–1622 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hwang, W. L. et al. Single-nucleus and spatial transcriptomics of archival pancreatic cancer reveals multi-compartment reprogramming after neoadjuvant treatment. Preprint at bioRxiv https://doi.org/10.1101/2020.08.25.267336 (2020).

  8. Chen, J. et al. Single-molecule dynamics of enhanceosome assembly in embryonic stem cells. Cell 156, 1274–1285 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu, Z. & Tjian, R. Visualizing transcription factor dynamics in living cells. J. Cell Biol. 217, 1181–1191 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rabani, M. et al. High-resolution sequencing and modeling identifies distinct dynamic RNA regulatory strategies. Cell 159, 1698–1710 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jovanovic, M. et al. Immunogenetics. Dynamic profiling of the protein life cycle in response to pathogens. Science 347, 1259038 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Rabani, M., Pieper, L., Chew, G.-L. & Schier, A. F. A massively parallel reporter assay of 3′ UTR sequences identifies in vivo rules for mRNA degradation. Mol. Cell 68, 1083–1094 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Gerlach, J. P. et al. Combined quantification of intracellular (phospho-)proteins and transcriptomics from fixed single cells. Sci. Rep. 9, 1469 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Reimegård, J. et al. A combined approach for single-cell mRNA and intracellular protein expression analysis. Commun. Biol. 4, 624 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Katzenelenbogen, Y. et al. Coupled scRNA-seq and intracellular protein activity reveal an immunosuppressive role of TREM2 in cancer. Cell 182, 872–885 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Rivello, F. et al. Single-cell intracellular epitope and transcript detection revealing signal transduction dynamics. Preprint at bioRxiv https://doi.org/10.1101/2020.12.02.408120 (2020).

  19. Mimitou, E. P. et al. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-00927-2 (2021).

  20. Mimitou, E. P. et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat. Methods 16, 409–412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Frangieh, C. J. et al. Multimodal pooled Perturb-CITE-seq screens in patient models define mechanisms of cancer immune evasion. Nat. Genet. 53, 332–341 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, Y. et al. Multiplexed in situ protein imaging using DNA-barcoded antibodies with extended hybridization chain reactions. Preprint at bioRxiv https://doi.org/10.1101/274456 (2018).

  23. Weichert, W. et al. High expression of RelA/p65 is associated with activation of nuclear factor-κB-dependent signaling in pancreatic cancer and marks a patient population with poor prognosis. Br. J. Cancer 97, 523–530 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lehmann, A. et al. High class I HDAC activity and expression are associated with RelA/p65 activation in pancreatic cancer in vitro and in vivo. BMC Cancer 9, 395 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Yang, S.-H. et al. Nuclear expression of glioma-associated oncogene homolog 1 and nuclear factor-κB is associated with a poor prognosis of pancreatic cancer. Oncology 85, 86–94 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Kim, H. D., Shay, T., O’Shea, E. K. & Regev, A. Transcriptional regulatory circuits: predicting numbers from alphabets. Science 325, 429–432 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Buccitelli, C. & Selbach, M. mRNAs, proteins and the emerging principles of gene expression control. Nat. Rev. Genet. 21, 630–644 (2020).

    Article  CAS  PubMed  Google Scholar 

  29. Lee, T. I. & Young, R. A. Transcriptional regulation and its misregulation in disease. Cell 152, 1237–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nelson, D. E. et al. Oscillations in NF-κB signaling control the dynamics of gene expression. Science 306, 704–708 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Hafner, A. et al. p53 pulses lead to distinct patterns of gene expression albeit similar DNA-binding dynamics. Nat. Struct. Mol. Biol. 24, 840–847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. O’Neill, L. A. J. & Kaltschmidt, C. NF-κB: a crucial transcription factor for glial and neuronal cell function. Trends Neurosci. 20, 252–258 (1997).

    Article  PubMed  Google Scholar 

  33. Spiegel, I. et al. Npas4 regulates excitatory–inhibitory balance within neural circuits through cell-type-specific gene programs. Cell 157, 1216–1229 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hu, P. et al. Dissecting cell-type composition and activity-dependent transcriptional state in mammalian brains by massively parallel single-nucleus RNA-seq. Mol. Cell 68, 1006–1015 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hrvatin, S. et al. Single-cell analysis of experience-dependent transcriptomic states in the mouse visual cortex. Nat. Neurosci. 21, 120–129 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Yap, E.-L. et al. Bidirectional perisomatic inhibitory plasticity of a Fos neuronal network. Nature 590, 115–121 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Sigal, A. et al. Dynamic proteomics in individual human cells uncovers widespread cell-cycle dependence of nuclear proteins. Nat. Methods 3, 525–531 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Purvis, J. E. et al. p53 dynamics control cell fate. Science 336, 1440–1444 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gaublomme, J. T. et al. Nuclei multiplexing with barcoded antibodies for single-nucleus genomics. Nat. Commun. 10, 2907 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. McKinley, K. L. & Cheeseman, I. M. Large-scale analysis of CRISPR/Cas9 cell-cycle knockouts reveals the diversity of p53-dependent responses to cell-cycle defects. Dev. Cell 40, 405–420 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Feldman, D. et al. Optical pooled screens in human cells. Cell 179, 787–799 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rosenberg, A. B. et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360, 176–182 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Saka, S. K. et al. Immuno-SABER enables highly multiplexed and amplified protein imaging in tissues. Nat. Biotechnol. 37, 1080–1090 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Aitchison, J. Measures of location of compositional data sets. Math. Geol. 21, 787–790 (1989).

    Article  Google Scholar 

  45. McGinnis, C. S. et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat. Methods 16, 619–626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Vezzani, A., French, J., Bartfai, T. & Baram, T. Z. The role of inflammation in epilepsy. Nat. Rev. Neurol. 7, 31–40 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Lévesque, M. & Avoli, M. The kainic acid model of temporal lobe epilepsy. Neurosci. Biobehav. Rev. 37, 2887–2899 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Lerea, L. S. & McNamara, J. O. Ionotropic glutamate receptor subtypes activate c-Fos transcription by distinct calcium-requiring intracellular signaling pathways. Neuron 10, 31–41 (1993).

    Article  CAS  PubMed  Google Scholar 

  49. Shen, W., Zhang, C. & Zhang, G. Nuclear factor κB activation is mediated by NMDA and non-NMDA receptor and L-type voltage-gated Ca2+ channel following severe global ischemia in rat hippocampus. Brain Res. 933, 23–30 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Kaltschmidt, B., Widera, D. & Kaltschmidt, C. Signaling via NF-κB in the nervous system. Biochim. Biophys. Acta 1745, 287–299 (2005).

    Article  CAS  PubMed  Google Scholar 

  51. Dammer, E. B. et al. Neuron enriched nuclear proteome isolated from human brain. J. Proteome Res. 12, 3193–3206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kierdorf, K. et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 16, 273–280 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Buus, T. B. et al. Improving oligo-conjugated antibody signal in multimodal single-cell analysis. eLife 10, e61973 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Habib, N. et al. Disease-associated astrocytes in Alzheimer’s disease and aging. Nat. Neurosci. 23, 701–706 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Saunders, A. et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell 174, 1015–1030 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Fleming, S. J., Marioni, J. C. & Babadi, M. CellBender remove-background: a deep generative model for unsupervised removal of background noise from scRNA-seq datasets. Preprint at bioRxiv https://doi.org/10.1101/791699 (2019).

  57. Morgan, J. I., Cohen, D. R., Hempstead, J. L. & Curran, T. Mapping patterns of c-Fos expression in the central nervous system after seizure. Science 237, 192–197 (1987).

    Article  CAS  PubMed  Google Scholar 

  58. Bergeron, D., Pal, G., Beaulieu, Y. B., Chabot, B. & Bachand, F. Regulated intron retention and nuclear pre-mRNA decay contribute to PABPN1 autoregulation. Mol. Cell Biol. 35, 2503–2517 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hughes, T. K. et al. Second-strand synthesis-based massively parallel scRNA-seq reveals cellular states and molecular features of human inflammatory skin pathologies. Immunity 53, 878–894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Crotti, A. et al. Mutant Huntingtin promotes autonomous microglia activation via myeloid lineage-determining factors. Nat. Neurosci. 17, 513–521 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lind, D., Franken, S., Kappler, J., Jankowski, J. & Schilling, K. Characterization of the neuronal marker NeuN as a multiply phosphorylated antigen with discrete subcellular localization. J. Neurosci. Res. 79, 295–302 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. Stein, B. et al. Cross-coupling of the NF-κB p65 and Fos/Jun transcription factors produces potentiated biological function. EMBO J. 12, 3879–3891 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Fujioka, S. et al. NF-κB and AP-1 connection: mechanism of NF-κB-dependent regulation of AP-1 activity. Mol. Cell Biol. 24, 7806–7819 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Cleary, B., Cong, L., Cheung, A., Lander, E. S. & Regev, A. Efficient generation of transcriptomic profiles by random composite measurements. Cell 171, 1424–1436 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Fernandez-Albert, J. et al. Immediate and deferred epigenomic signatures of in vivo neuronal activation in mouse hippocampus. Nat. Neurosci. 22, 1718–1730 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Gupta, S., Campbell, D., Dérijard, B. & Davis, R. J. Transcription factor ATF2 regulation by the JNK signal transduction pathway. Science 267, 389–393 (1995).

    Article  CAS  PubMed  Google Scholar 

  67. Vierbuchen, T. et al. AP-1 transcription factors and the BAF complex mediate signal-dependent enhancer selection. Mol. Cell 68, 1067–1082 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Strauss, K. I. et al. Prolonged cyclooxygenase-2 induction in neurons and glia following traumatic brain injury in the rat. J. Neurotrauma 17, 695–711 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Gayoso, A. et al. Joint probabilistic modeling of single-cell multi-omic data with totalVI. Nat. Methods 18, 272–282 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hartmann, F. J. et al. Single-cell metabolic profiling of human cytotoxic T cells. Nat. Biotechnol. 39, 186–197 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Qiu, Q. et al. Massively parallel and time-resolved RNA sequencing in single cells with scNT-seq. Nat. Methods 17, 991–1001 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Boon, R., Silveira, G. G. & Mostoslavsky, R. Nuclear metabolism and the regulation of the epigenome. Nat. Metab. 2, 1190–1203 (2020).

    Article  CAS  PubMed  Google Scholar 

  73. Ma, S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Biancalani, T. et al. Deep learning and alignment of spatially-resolved whole transcriptomes of single cells in the mouse brain with Tangram. Preprint at bioRxiv https://doi.org/10.1101/2020.08.29.272831 (2020).

  75. Amamoto, R. et al. FIN-seq: transcriptional profiling of specific cell types from frozen archived tissue of the human central nervous system. Nucleic Acids Res. 48, e4 (2020).

    CAS  PubMed  Google Scholar 

  76. Racine, R. J. Modification of seizure activity by electrical stimulation: II. Motor seizure. Electroencephalogr. Clin. Neurophysiol. 32, 281–294 (1972).

    Article  CAS  PubMed  Google Scholar 

  77. Li, B. et al. Cumulus provides cloud-based data analysis for large-scale single-cell and single-nucleus RNA-seq. Nat. Methods 17, 793–798 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Bakken, T. E. et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS ONE 13, e0209648 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Stoeckius, M. et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. McInnes, L. et al. UMAP: Uniform Manifold Approximation and Projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  83. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Schmid-Burgk and I. Cheeseman for the HeLa p65–mNeonGreen reporter line, L. Gaffney for assistance with figure preparation, P. Thakore for coining the acronym inCITE-seq, A. Rubin for critical feedback on the manuscript, C. McGinnis for helpful sharing of data, the Broad Institute Flow Cytometry Core facility, and all members of the Regev laboratory for helpful discussions. This research was supported by NIH/NHGRI CEGS grant 5RM1 HG006193. A.R. was a Howard Hughes Medical Institute Investigator (until 31 July 2020). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

H.C. conceived and designed the study with guidance from A.R. C.N.P. designed and performed mouse experiments with guidance from D.A. H.C. and E.M.M. developed and performed inCITE-seq experiments, with buffer optimization input from F.C. and early-stage experimental support from J.W. C.N.P. and E.M.M. conducted immunohistochemistry. D.P. conducted 10x experiments and constructed sequencing libraries. B.Z.Y. conjugated inCITE antibodies. H.C. analyzed and interpreted data with help from E.H. on cis regulatory motif enrichment analysis and supervision from A.R. A.R. provided project oversight and funding. H.C. and A.R. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Hattie Chung or Aviv Regev.

Ethics declarations

Competing interests

A.R. is a founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas Therapeutics and, until 31 August 2020, was an SAB member of Syros Pharmaceuticals, Neogene Therapeutics, Asimov and Thermo Fisher Scientific. From 1 August 2020, A.R. is an employee of Genentech. From May 2021, D.P. is an employee of Genentech. B.Z.Y. was formerly an employee of BioLegend and is now an employee of Spatial Genomics. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Methods thanks Dominic Grun, Bing Ren and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Lei Tang was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Optimization of intranuclear antibody staining in HeLa cells.

a. Nuclear p65 levels change after TNFα treatment, while total p65 in cells remains unchanged. Distribution of p65-mNeonGreen reporter fluorescence (x axis; % mode of singlet nuclei, y axis) measured by flow cytometry of nuclei (solid line) vs. cells (dashed line) from untreated (‘NT’, blue) or TNFα treated cells (red). b. Flow cytometry distinguishes p65-mNeonGreen signals across mixtures of NT and TNFα. Top: Flow cytometry measures of mNeonGreenhigh fraction (x axis) match the input fraction of TNFα nuclei (x axis). Bottom: Corresponding high (red) and low (blue) mNeonGreen distributions. c. Immunofluorescence of nuclei smeared onto a slide after intranuclear p65 stain in suspension, showing complete antibody diffusion into the nucleus; representative of 3 experiments. Scale: 100 µm. d,e. Comparing antibody- and fluorescence reporter-derived p65 levels. Antibody (from Alexa Fluor 647 secondary, y axis) and mNeonGreen (x axis) signal of p65 in an equal mixture of NT and TNFα stimulated nuclei. Histograms: marginal distributions. d. Agreement between unconjugated p65 antibody and mNeonGreen signal. e. No relationship between DNA-conjugated p65inCITE-Ab and mNeonGreen signal using standard intranuclear staining buffer (pre-optimization). f. Relation between nuclei hashtag oligonucleotide (HTO; x axis) counts and p65 antibody-derived tag (ADT; y axis) counts, shown across 10,014 NT and TNFα nuclei, colored by the number of RNA UMIs. Top left: Pearson R2 and associated P-value (two-sided t-test). To control for this relation, we normalize protein ADT counts by nuclei HTO counts (Methods). g. Comparing RNA complexity from inCITE-seq (fixed HeLa nuclei) and MULTI-seq (unfixed HEK nuclei, from McGinnis et al.45) by the distribution of the number of detected transcripts (UMIs; top) and genes (bottom). h. Low correlation between p65 protein (y axis, nCLR) and RELA RNA levels (x axis, log normalized), with Pearson R2 and associated P-value (two-sided t-test). Dots: nuclei colored by treatment (NT, blue; TNFα, red). i. Dynamics of gene expression after LPS stimulation in mouse dendritic cells, from Rabani et al.10, measured across time (x axis). Relative expression to steady state, t0 (y axis): pre-mRNA precursor (blue) and mRNA (red) for total (solid) vs. 4sU labeled (dashed) RNA, shown for Rela (top) and Nfkbia (bottom), a p65 target as in Fig. 1e.

Source data

Extended Data Fig. 2 Flow cytometry of inCITE targets on nuclei or cells extracted from frozen mouse hippocampus.

Flow cytometry of nuclei populations from the mouse hippocampus after intranuclear stains with inCITE antibodies, followed by Alexa Fluor 647-conjugated secondary stain: NeuN in PBS (a), PU.1 in PBS (b), p65 in kainic acid (KA) (d), and c-Fos in PBS (e) and KA (f) treated mice. Axes show fluorescence signal (x axis) and side scatter (y axis) of singlet nuclei (dots); histograms show marginal distributions. Oval gates show NeuNhigh (a, 58.3%), PU.1high (b, <3%), p65high (d, 55.2%), c-Foshigh (0.21% in PBS (e), and 48.7% after KA treatment (f)). c. Right: Distribution of PU.1 in microglia (CD11b+ CX3CR1+, red), CD4+ cells (blue) and isotype (gray) cells measured by flow cytometry (left and middle panels) after simultaneous surface protein and intracellular protein stains (Methods).

Extended Data Fig. 3 Antibody signal varies across concentration regimes.

Antibody stains of the mouse hippocampus (extracted nuclei or in situ) with inCITE antibodies across a wide range of dilutions, targeting NeuN in PBS (a,e), PU.1 in PBS (b,f), p65 in kainic acid (c,g), and c-Fos in kainic acid (d,h) treated mice. Antibody-derived fluorescence measured by Alexa Fluor 647-conjugated secondary antibody stain. a-d. Histograms are normalized as % mode of nuclei singlets. Antibody dilutions are indicated to the right of each axis, with dilutions used for inCITE-seq in bold (NeuN 1:500, PU.1 1:200, p65 1:400, c-Fos 1:400). e-h. In situ immunofluorescence of frozen mouse hippocampus with inCITE antibodies across different dilutions, matching the concentrations used in flow cytometry; representative of 2 independently conducted experiments. Scale bars, 100 µm.

Extended Data Fig. 4 Impact of tissue preparation on epitope detection by antibodies.

Comparing in situ immunofluorescence of antibody stains (followed by Alexa Fluor 647-conjugated secondary stain) in mouse hippocampus tissue that were immediately frozen (green box) or frozen after overnight fixation in 4% PFA (purple box, Methods) across a wide range of antibody dilutions. Images are representative of 2 independent experiments. a. NeuN in PBS. Biolegend NeuN antibody (clone 1B7) used for inCITE and Abcam NeuN antibody (clone EPR12763). b. PU.1 in PBS. Biolegend PU.1 antibody (clone 7C2C34) used for inCITE and Cell Signaling Technology PU.1 antibody (clone 9G7). c. p65 in KA. Biolegend p65 antibody (clone Poly6226) used for inCITE. d. c-Fos in KA treated mice. Biolegend c-Fos antibody (clone Poly6414) used for inCITE and Abcam c-Fos antibody (ab190289). Scale bars, 100 µm.

Extended Data Fig. 5 Comparing and combining single nucleus RNA profiles from inCITE-seq and snRNA-seq of mouse hippocampus.

a. Comparing the complexity of RNA profiles from inCITE-seq and standard snRNA-seq of the mouse hippocampus. Distributions (marginals) of the number of UMIs (x axis) and genes (y axis) from inCITE-seq (left), matching mouse hippocampus snRNA-seq in this study (middle), and previously published snRNA-seq (right). Scatter plot shows the density of individual nuclei (dots) calculated with a Gaussian kernel estimate. b,c. Major cell types from the adult mouse hippocampus identified from inCITE-seq RNA profiles alone. b. UMAP embedding of 24,444 single nucleus inCITE-seq RNA profiles (dots) colored by annotated cluster (number). c. Expression of marker genes (columns) used for annotating cell type clusters (rows), showing mean expression of log normalized counts (dot color) and proportion of expressing cells (dot size). d-j. Enhanced cell type distinctions and annotation by combining RNA profiles from inCITE-seq and snRNA-seq. Joint UMAP embedding of 22,260 inCITE-seq and 15,507 snRNA-seq RNA profiles (dots) colored by unsupervised leiden clusters or subcluster of leiden group 4 (numbers) (Methods). e. Distribution of mitochondrial fraction of total gene content (y axis, left) and total transcript counts (y axis, right) in each leiden cluster or subcluster of leiden group 4 (x axis, both). Asterisks indicate cluster 15 (n = 327 nuclei) and subcluster 4,3 (n = 179 nuclei) that were removed for high mitochondrial content and for low RNA complexity, respectively. f-h. UMAP embedding as in Fig. 2d colored by doublets that were removed from subsequent analyses (n = 3,059 doublets, (f)), batch and assay (g), or condition (h). i. Percent of nuclei (y axis) from each batch/assay (color) in each cluster (x axis). j. Mean expression of log normalized counts (dot color) and proportion of expressing cells (dot size) of marker genes (columns) used for annotating cell type clusters in d (rows).

Source data

Extended Data Fig. 6 Protein levels by inCITE-seq batch (replicate).

a-d. Distribution of protein levels (x axis, nCLR) shown as kernel density estimates of NeuN (a), PU.1 (b), p65 (c), or c-Fos (d) in each batch (top: batch 1; bottom: batch 2) in biologically relevant subsets as foreground (color) and appropriate background set of nuclei (grey). Dashed line: Batch-specific threshold used to partition protein level as high vs. low. e-i. Density distribution of (e) nucleus hashtag counts (x axis, HTOs) or (f-i) antibody-derived tags (x axis, ADTs) of inCITE target proteins, colored by batch (batch 1, gray; batch 2, blue).

Source data

Extended Data Fig. 7 Protein effects on global gene expression.

a. Relation between unspliced pre-mRNA expression of Rbfox3 and nuclear protein levels of NeuN. Distribution of pre-mRNA levels (Z score of log-normalized counts, y axis) in nuclei with high or low levels of NeuN (x axis) after PBS (gray) or KA (green) treatment (NeuN thresholds in Extended Data Fig. 6). Boxplots show the median (centre line), box bounds represent first and third quartiles, and whiskers span from each quartile to the minimum or the maximum (1.5 interquartile range below 25% or above 75% quartiles). Dots correspond to 227 individual nuclei with non-zero pre-mRNA levels measured across n = 2 biologically independent samples. Significance, from left: P = 5*10−15, P = 9*10−5 two-sided Mann-Whitney test. NS – not significant. b. Functional gene sets enriched in TF associated genes. Enrichment (-log10(P-value), x axis, hypergeometric test) of Gene Ontology (GO) terms (y axis) in genes significantly associated (from top to bottom) with p65 (33 genes), PU.1 (13 genes), and c-Fos (10 genes). c. Genes associated with NeuN. Effect size (x axis) and associated significance (y axis, -log10(P-value)) for the association of each gene (dots) with NeuN by a model of gene expression as a linear combination of the four inCITE-seq target proteins after regressing out treatment and cell type (Methods). Select genes are labeled. Colored dots: Benjamini-Hochberg FDR < 5%.

Source data

Extended Data Fig. 8 Genes and modules associated with TFs within excitatory (EX) neurons.

a. Genes associated with protein-protein pairs in the interaction model, identified by modeling gene expression across excitatory neurons as a linear combination of individual proteins and their pairwise interactions after regressing out treatment. Effect size (x axis) and significance (y axis, -log10(P-value)) for DEGs (dots) associated with each protein-protein interaction term: p65 and c-Fos (left), c-Fos and NeuN (middle), and p65 and NeuN (right). Select genes are labeled. Colored dots: Benjamini-Hochberg FDR < 5%. b. Pearson correlation coefficient (red/blue colorbar) of pairwise gene expression profiles (rows and columns) significantly (FDR < 5%) associated positively (purple) or negatively (green), with c-Fos (additive model), p65 (additive model), or c-Fos*p65 (interaction model), ordered by hierarchical clustering. Top bars: Effect size of each protein or protein-protein pair. c. Treatment effect on gene programs. Program scores (y axis) for 5 EX programs (in Fig. 4f) of 15,226 individual nuclei (dots) from PBS or KA treated mice (x axis) measured across 2 biologically independent experiments. Boxplots show the median (centre line), box bounds represent first and third quartiles, and whiskers span from each quartile to the minimum or the maximum (1.5 interquartile range below 25% or above 75% quartiles). Significance, from left: P= 0.049, P= 2.7*10−271, P = 2.2*10−199, P = 6.1*10−7, two-sided Mann-Whitney test. NS – not significant.

Source data

Extended Data Fig. 9 Treatment-dependent cis-regulatory elements and TF-associated genes.

a-c. Prediction of co-regulatory patterns by TF motif enrichment in DEGs associated with c-Fos or p65 (additive model), or their interaction c-Fos*p65 (interaction model). a,b. Significance (-log10(P-value), y axis) and rank order (x axis) of TF motifs (dots) enriched in enhancers of DEGs associated with each protein (additive model) or protein-protein (interaction model) term in excitatory neurons, using enhancers of PBS (a) or KA (b) treated sample as background. Black: significant motifs (P< 10−3, hypergeometric test); gray: not significant. c. TF motif enrichment (columns; dot size, -log10(P-value)) and proportion of excitatory neuron nuclei expressing the RNA (color) of significant TFs (rows) in the enhancers of c-Fos (additive model), p65 (additive model), or c-Fos*p65 (interaction model) DEGs, compared to other enhancers within the KA treated sample. d. Treatment-dependence of gene association with c-Fos and p65. Global effect size of genes (dots) associated with c-Fos (left) and p65 (right), after PBS (x axis) or KA treatment (y axis) (Methods). Colored dots: genes with significant coefficients (Benjamini-Hochberg FDR < 5%) in PBS (gray), KA (green), or both (black). Select genes are labeled. Bottom right: linear correlation R2 and associated P value (two-sided t-test).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chung, H., Parkhurst, C.N., Magee, E.M. et al. Joint single-cell measurements of nuclear proteins and RNA in vivo. Nat Methods 18, 1204–1212 (2021). https://doi.org/10.1038/s41592-021-01278-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01278-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing